PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in...

download PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

of 21

Transcript of PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in...

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    1/21

    This Provisional PDF corresponds to the article as it appeared upon acceptance. Fully formattedPDF and full text (HTML) versions will be made available soon.

    PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidenceand implications in clinical outcome

    BMC Cancer 2012, 12:514 doi:10.1186/1471-2407-12-514

    Purificacion Estevez-Garcia ([email protected])Angel Castao ([email protected])Ana C Martin ([email protected])

    Fernando Lopez-Rios ([email protected])Joaquin Iglesias ([email protected])

    Sandra Muoz-Galvn ([email protected])Iker Lopez-Calderero ([email protected])

    Sonia Molina-Pinelo ([email protected])Maria D Pastor ([email protected])

    Amancio Carnero ([email protected])Luis Paz-Ares ([email protected])

    Rocio Garcia-Carbonero ([email protected])

    ISSN 1471-2407

    Article type Research article

    Submission date 22 June 2012

    Acceptance date 28 October 2012

    Publication date 12 November 2012

    Article URL http://www.biomedcentral.com/1471-2407/12/514

    Like all articles in BMC journals, this peer-reviewed article can be downloaded, printed anddistributed freely for any purposes (see copyright notice below).

    Articles in BMC journals are listed in PubMed and archived at PubMed Central.

    For information about publishing your research in BMC journals or any BioMed Central journal, go to

    http://www.biomedcentral.com/info/authors/

    BMC Cancer

    2012 Estevez-Garcia et al.This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0),

    which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

    mailto:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]://www.biomedcentral.com/1471-2407/12/514http://www.biomedcentral.com/info/authors/http://creativecommons.org/licenses/by/2.0http://creativecommons.org/licenses/by/2.0http://www.biomedcentral.com/info/authors/http://www.biomedcentral.com/1471-2407/12/514mailto:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]
  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    2/21

    PDGFR/ and VEGFR2 polymorphisms in

    colorectal cancer: incidence and implications inclinical outcome

    Purificacion Estevez-Garcia1,2

    Email: [email protected]

    Angel Castao3

    Email: [email protected]

    Ana C Martin4

    Email: [email protected]

    Fernando Lopez-Rios5

    Email: [email protected]

    Joaquin Iglesias4

    Email: [email protected]

    Sandra Muoz-Galvn1

    Email: [email protected]

    Iker Lopez-Calderero1,2

    Email: [email protected]

    Sonia Molina-Pinelo1

    Email: [email protected]

    Maria D Pastor1

    Email: [email protected]

    Amancio Carnero1

    Email: [email protected]

    Luis Paz-Ares1,2

    Email: [email protected]

    Rocio Garcia-Carbonero1,2,*

    Email: [email protected]

    1Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de

    Sevilla), Sevilla, Spain

    2Medical Oncology Department, Hospital Universitario Virgen del Rocio,

    Sevilla, Spain

    3 Pathology Department, Hospital de Fuenlabrada, Madrid, Spain

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    3/21

    4Bionostra Aplicaciones Biotecnolgicas, S.L.U., Madrid, Spain

    5Pathology Department, Centro Integral Oncolgico Clara Campal, Madrid,

    Spain

    *

    Corresponding author. Medical Oncology Department, Hospital UniversitarioVirgen del Rocio, Sevilla, Spain

    Abstract

    Background

    Angiogenesis plays an essential role in tumor growth and metastasis, and is a major target in

    cancer therapy. VEGFR and PDGFR are key players involved in this process. The purpose of

    this study was to assess the incidence of genetic variants in these receptors and its potential

    clinical implications in colorectal cancer (CRC).

    Methods

    VEGFR2, PDGFR and PDGFR mutations were evaluated by sequencing their tyrosine

    kinase domains in 8 CRC cell lines and in 92 samples of patients with CRC. Correlations

    with clinicopathological features and survival were analyzed.

    Results

    Four SNPs were identified, three in PDGFR [exon 12 (A12): c.1701A>G; exon 13 (A13):c.1809G>A; and exon 17 (A17): c.2439+58C>A] and one in PDGFR [exon 19 (B19):

    c.2601A>G]. SNP B19, identified in 58% of tumor samples and in 4 cell lines (LS174T,

    LS180, SW48, COLO205), was associated with higher PDGFR and pPDGFR protein levels.

    Consistent with this observation, 5-year survival was greater for patients with PDGFR B19

    wild type tumors (AA) than for those harboring the G-allele genotype (GA or GG) (51% vs

    17%; p=0.073). Multivariate analysis confirmed SNP B19 (p=0.029) was a significant

    prognostic factor for survival, independent of age (p=0.060) or TNM stage (pG SNP is commonly encountered in CRC patients and isassociated with increased pathway activation and poorer survival. Implications regarding its

    potential influence in response to PDGFR-targeted agents remain to be elucidated.

    Keywords

    VEGFR, PDGFR, SNP, Colorectal cancer, Angiogenesis, Prognosis

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    4/21

    Background

    Colorectal cancer (CRC) is the third most common tumour in the world, with over 1.2 million

    new cases diagnosed every year, and is responsible for about 8% of cancer related deaths [1].

    Approximately one third of patients present metastatic disease at diagnosis, and about 40% of

    those with early-stage tumors will eventually relapse at some point over the course of the

    disease [2,3]. Although prognosis has greatly improved over the past decades due to

    significant surgical and medical advances, once the tumor has progressed beyond surgical

    resectability, the disease is essentially incurable and median survival ranges from 14 to 24

    months with best available systemic therapy [4]. Development of new more effective agents

    is thus actively pursued.

    Angiogenesis has become a major target in colorectal cancer therapy. Bevacizumab, a

    humanized monoclonal antibody against the vascular endothelial growth factor A (VEGF-A),

    was the first antiangiogenic agent to demonstrate efficacy in CRC. In the pivotal study by

    Hurwitz et al., the addition of this agent to irinotecan-based combination cytotoxic therapysignificantly improved survival compared to irinotecan-based chemotherapy alone in patients

    with advanced CRC [5]. Subsequently, bevacizumab has been tested in combination with

    other chemotherapy regimens with more modest results [3,4]. More recently, a benefit in

    survival has been also reported in patients with advanced CRC with two new promising

    antiangiogenic drugs: aflibercept (a VEGF trap) in combination with FOLFIRI (folinic acid,

    5-fluoruracil and irinotecan) following progression to oxaliplatin-based therapy [6], and

    regorafenib (a novel tyrosine kinase inhibitor targeting VEGFR, PDGFR, FGFR, RET, KIT

    and TIE2) as single-agent therapy in patients who had progressed to all standard therapies

    [7]. These results clearly illustrate angiogenesis inhibition is to play a major role in the

    management of this disease.

    Angiogenesis is a highly controlled process under physiological conditions, such as

    embryonal development, postnatal growth and wound healing, but is also a critical driver of

    tumor growth and progression [8]. It is tightly regulated by a complex equilibrium among

    different pro- and antiangiogenic factors secreted both by tumor cells and by cells of the

    tumor microenvironment (pericytes, endothelial, mesenchymal or immune cells). VEGF and

    their receptors represent one of the best validated pathways involved in angiogenesis [9,10].

    VEGF stimulates both proliferation and migration of endothelial cells, enhances

    microvascular permeability, and is essential for revascularization during tumor formation. It

    is commonly over-expressed in human tumors, and this is often associated with increased

    vascular density and more aggressive clinical behavior. VEGF-A and its main receptor,

    VEGFR2/KDR, are key members of this family and common targets of antiangiogenic agents[11,12].

    Platelet-derived growth factor (PDGF) and their receptors (PDGFR-, PDGFR- and

    PDGFR-) play also a critical role in angiogenesis regulation by exerting important control

    functions in mesenchymal cells during development [13]. PDGF is expressed by endothelial

    cells and acts in a paracrine manner by recruiting PDGFR-expressing cells, such as pericytes

    and smooth muscle cells, to the developing vessels, thus improving pericyte coverage and

    vessel function. PDGF signaling promotes cell migration, survival and proliferation and

    indirectly regulates angiogenesis by inducing VEGF transcription and secretion [10,13,14].

    Mutations involving up-regulation of PDGF and/or PDGFR, as well as PDGFR-dependent

    growth stimulation, have been documented in a number of solid tumors and hematological

    malignancies, suggesting a likely role of this pathway in carcinogenesis [10,15]. Moreover,

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    5/21

    agents antagonizing PDGFR-mediated signaling have also demonstrated antineoplastic

    activity in preclinical models and in clinical trials, including some conducted in patients with

    CRC (i.e. regorafenib) [7]. Nevertheless, several other drugs also targeting these pathways

    (i.e. sunitinib, sorafenib) [16,17] have failed to prove a significant positive impact on the

    outcome of patients with CRC. The biological grounds for these discordant results are not

    well understood.

    Therefore, and in spite of their undeniable success, only a small proportion of patients do

    actually benefit from antiangiogenic agents, and reliable tools to prospectively identify which

    patients are more likely to benefit are scarce. In this scenario, efforts to unravel the intricate

    molecular pathways governing tumor angiogenesis are certainly needed for progress to be

    made. In the present study, we sought to evaluate the incidence of genetic polymorphisms of

    some of the key players of angiogenesis, such as VEGFR-2, PDGFR-A and PDGFR-B, and

    their potential influence in CRC biology. With this purpose we sequenced the tyrosine kinase

    domains of these receptors in 8 CRC cell lines (T84, LOVO, LS174T, HT29, LS180, SW48,

    SW480, COLO205) and in 92 tumor samples of patients with colorectal adenocarcinoma.

    Correlations of encountered genetic variables with protein expression in cell lines, as well aswith clinicopathological features and survival of these patients were also analyzed to assess

    their potential biological and clinical implications.

    Methods

    Laboratory Procedures

    CRC cell lines

    Eight human CRC cell lines (T84, LOVO, LS174T, HT 29, LS180, SW48, SW480 andCOLO205) were selected and purchased from the European Collection of Cell Cultures

    (ECACC). They were representative of patients with different gender, age and tumor stage.

    Cell culture

    Each cell line was grown in conditions of temperature, humidity, O2 and CO2 levels, culture

    medium and supplements according to providers instructions. Once they reached confluence

    in monolayer DNA extraction was performed. The total DNA yield was determined using a

    Nanodrop ND-1000 spectrophotometer (Nanodrop Tech, DE, USA).

    DNA isolation from human tumor samples and culture cells

    Formalin-fixed paraffin-embedded tissues from the 92 selected CRC patients were provided

    by the Pathology Departments of the corresponding institutions. Samples were mainly

    obtained from the primary tumor (96%), either by surgical (87%) or endoscopic procedures.

    Three tissue sections of each tumor were first deparaffinized and rehydrated by serial passes

    in D-Limoneno (Histo-Clear, National Diagnostic, Atlanta, GA, USA) and ethanol (100%).

    Then, DNA isolation from both human tumor tissue samples and culture cells was performed

    with the REAL pure genomic DNA extraction kit (Durviz, Valencia, Spain) according to the

    manufacturers instructions and then then purified using ion exchange columns (QIAGEN

    Miniprep kit Cat. No. 27106). The total DNA yield was determined using a Nanodrop ND-1000 spectrophotometer (Nanodrop Tech, DE, USA).

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    6/21

    Genotyping

    Public databases including National Center for Biotechnology Information (NCBI)

    (http://www.ncbi.nlm.nih.gov), University of California Santa Cruz (UCSC) Genome

    Bioinformatics (http://genome.ucsc.edu) and Ensembl Genome Browser

    (http://www.ensembl.org/index.html) were reviewed to obtain the haplotypes of the threegenes of interest and their reported genetic variants. The exomic regions corresponding to the

    tyrosine kinase domains, which were the regions with the highest probability of mutations,

    were then identified for each gene: exons 17 to 26 for VEGFR2, and exons 12 to 21 for

    PDGFR and PDGFR. Specific primers were designed to amplify these exons using expert

    software in order to minimize non-specific or erroneous amplifications and improve

    outcomes. Primers used in this study are described in Additional file 1: Table S1.

    Amplification of the tyrosine kinase domains in both CRC cell lines and tissue samples was

    performed by a polymerase chain reaction (PCR) method. Fifty nanograms of the genomic

    purified DNA were amplified in a PCR reaction containing 1.5 units of DNA polymerase

    EuroTAQ (Genycell Biotech Spain SL; Santa Fe, Granada, Spain), 1xEuroTaq buffer, 2.5

    mM Mg2+, 0.4 M forward and reverse primers, 80 M dNTPs (20 M each one), 1% DMSOand 1M betaine in a volume of 50 l. The PCR cycling conditions were as follows: initial

    denaturation at 94C for 5 minutes, 5 cycles at 94C for 1 minute, and annealing that began at

    67C for 45 seconds; this temperature was decreased 2C each cycle to 59C (67, 65, 63, 61,

    59) and then 45 seconds at 72C. This was followed by 35 cycles at 95C 1 minute, 55C for

    45 seconds and 72C for 45 seconds. The last step was a final extension cycle at 72C for 10

    minutes.

    DNA sequencing

    PCR products were first purified using the microClean kit (Microzone Ltd.; Haywards Heath,UK) or ExoSAP-IT for PCR Product Clean-Up USB (Affimetrix Inc; Santa Clara, CA,

    USA) for individual reactions or PERFORMADTV V396-Well Short Plates (Genycell

    Biotech Spain SL; Santa Fe, Granada, Spain) for 96 plate reactions. Direct bidirectional

    sequencing of the PCR products was done using BigDyeTerminator Cycle v3.1 Sequencing

    Kit (Applied Biosystems; Carlsbad, CA, USA) and ABI 3110 Genetic Analyser (Applied

    Biosystems) according to the manufacturers instructions. All fragments were double-strand

    sequenced a number of times, and genetic variations found were checked twice. Sequencing

    analysis was performed using Chromas Lite, Clustal W and DiAlign software.

    Analysis of protein expression

    Cells were washed twice in 1x PBS, pelleted for 30 seconds at 14000x g and lysed in lysis

    buffer (TrisHCl pH 7.5 50 mM, NP40 1%, glycerol 10%, NaCl 150mM, complete protease

    inhibitor cocktail, 2 mM; Roche). After centrifugation, supernatant protein extracts were

    aliquoted and stored at 80C until use. The amount of protein was determined by Bradford

    assay using BSA (bovine serum albumin) as a standard. The appropriate protein quantity was

    dissolved in Laemli buffer (TrisHCl pH 6.8 62.5mM, glycerol 10%, SDS 1%, 2-mercapto

    ethanol 5%, bromphenol blue 0.0025%) and the proteins were separated in SDS-PAGE gels

    (12%) before they were blotted onto Nitrocellulose Transfer membrane (Whatman -

    Protrans). Primary antibodies employed were: p-PDGFR- (Tyr1021)-R 1:400 (Santa

    Cruz#sc-12909-R), PDGFR- 1:500 (Santa Cruz#sc-339), tubulin 1:10000 (SigmaT6557).

    The secondary antibodies used were goat anti-rabbit Alexa Fluor 680 1:5000 (Invitrogen A21057) and donkey anti-mouse IRDye 800CW 1:5000 (Rockland Inc.605-731-002).

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    7/21

    CRC study population, tumor samples and data collection

    Patients that met the following inclusion criteria were selected for the present study: (1)

    histologically confirmed diagnosis of primary CRC; (2) adequate clinical data recorded in

    medical charts; (3) adequate tissue specimen available for additional molecular assays (a

    proportion of tumor cells > 50% was required). Cases were reviewed according to apreviously designed protocol which included the following clinical data: age, sex, date of

    diagnosis, baseline carcinoembryonic antigen (CEA) plasma levels, primary tumor location,

    TNM stage [18], histological type, tumor differentiation, surgical treatment (type and

    outcome of surgery), chemotherapy (adjuvant or for advanced disease, regimen used),

    radiotherapy (neoadjuvant, adjuvant or palliative), date of last visit or death and cause of

    death. The study protocol was approved by the institutional review boards of participating

    centers.

    Main characteristics of the 92 included patients are summarized in Table 1 and are

    representative of a standard CRC population. The median age was 68 years, 63% were male

    and 40% presented advanced disease at diagnosis. The great majority had conventional

    adenocarcinomas (86%) and only 13% were poorly differentiated tumors. Cancer specific

    therapy is outlined in Additional file 1: Table S2. Patients with early stage disease (I-III)

    underwent primary tumor surgery with curative intent. Adjuvant fluoropyrimidine-based

    chemotherapy with or without oxaliplatin was indicated in patients with high risk stage II or

    stage III CRC following surgical resection. Neoadjuvant or adjuvant radiotherapy was added

    in stage II-III patients with rectum primaries. Patients with advanced stage IV disease were

    managed primarily with systemic chemotherapy that included oxaliplatin- (44%) or

    irinotecan-based (13%) combination regimens or fluoropyrimidines alone (3%). With a

    median follow-up of 31 months (range: 8 to 99 months), 59 patients (64%) had died due to

    disease progression or to complications of cancer therapy.

    Table 1Population and tumor samples characteristics

    N (%)

    Age (years)

    Median (range) 68 (4587)

    Gender

    Female 34 (37.0%)

    Male 58 (63.0%)

    Primary tumor location

    Right colon 27 (29.3%) Transverse colon 5 (5.4%)

    Left colon 9 (9.8%)

    Sigmoid colon 18 (19.6%)

    Recto sigmoid colon 14 (15.2%)

    Rectum 19 (20.7%)

    Histology

    Conventional adenocarcinoma 79 (85.9%)

    Mucinous or colloid adenocarcinoma 12 (13.0%)

    Signet ring cell adenocarcinoma 1 (1.1%)

    Tumor differentiation

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    8/21

    Well differentiated 22 (23.9%)

    Moderately differentiated 46 (50.0%)

    Poorly differentiated 12 (13.0%)

    TNM stage

    I 8 (8.7%) II 22 (23.9%)

    III 24 (26.1%)

    IV 37 (40.2%)

    Unknown 1 (1.1%)

    Baseline CEA (ng/mL)

    High 33 (35.9%)

    Normal 59 (64.1%)

    Median (range) 3 (013.318)

    TNM: Tumor, Node and Metastases; CEA: carcinoembryonic antigen

    Statistical analysis

    A minimum sample size of 80 patients was planned to be screened in case no mutations were

    to be encountered, as in such a case the probability of finding mutations in the general

    population was estimated to be very low ( 4.4%; =0.05, =0.80) and therefore non-

    clinically relevant. Considering an expected drop-out rate of about 10% (technical issues or

    others), 92 patients were finally selected for study entry. Descriptive statistics were used to

    characterize the most relevant clinical parameters. The association of categorical clinical or

    pathological features and mutation type was explored by the chi-squared test or Fishers exact

    test when appropriate. Overall survival (OS) was calculated from the time of histological

    diagnosis to the date of death (deaths due to surgical complications were censored). The

    Kaplan-Meier product limit method [19] was used to estimate OS, and differences observed

    among patient subgroups were assessed by the log rank test [20]. Multivariate analysis using

    the Cox proportional hazards model [21] was performed to assess the association between

    mutations and clinical outcome while adjusting for other potential confounding factors such

    as age, tumor stage, primary tumor location, CEA levels and tumor differentiation. P

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    9/21

    SNPs identified in CRC cell lines

    Both SNP A12 and SNP A17 were found in homozygosis in all CRC cell lines. PDGFR-A13

    SNP was present in heterozygosis in two cell lines (LS174T and LS180), and PDGFR-B19

    presented a SNP in heterozygosis in four of them (LS174T, LS180, SW48 and SW480).

    SNPs identified in CRC patient tumor samples

    PDGFR-A12 and PDGFR-A17 analysis was feasible in all tumor samples, and all of them

    showed the SNPs variants in homozygosis. PDGFR-A13 was successfully analyzed in 73

    cases (79%), being the SNP A13 detected in heterozygosis in 18% of analyzed samples (13

    patients). PDGFR-B19 complete analysis was achieved in 78 patients (85%), and the SNP

    B19 was found in 58% of evaluable samples (45 patients), both in homo- and heterozygosis

    (7 and 38 patients, respectively). Figure 1 illustrates DNA sequencing of PDGFR exon 12

    and PDGFR exon 19, showing SNPs identified in our population.

    Figure 1Electropherogram of a PDGFR sequence. 1a: Three examples of paraffin-embedded tumour tissue DNA sequence analysis of PDGFR exon wild type (Adenine), and

    SNP B19 in heterozygosis and homozygosis (A to G transition), respectively. 1b: DNA

    sequence of a paraffin-embedded tumour tissue sample presenting PDGFR exon 13 SNP in

    heterozygosis (G to A transition)

    Correlation of PDGFR and PDGFR genetic variants and

    clinicopathological features

    Distribution of SNPs A13 and B19 according to gender, age, baseline CEA levels, primary

    tumor location, histological type, TNM stage at diagnosis and tumor differentiation isdescribed in Table 2. The only observed correlations that were of borderline statistical

    significance were those found between SNP B19 and primary tumor location, and SNP A13

    and tumor differentiation. Indeed, the PDGFR B19 SNP was more commonly encountered

    among patients with colon primaries than in those with primary tumors located in the rectum

    (63.9% vs 35.3%; p=0.051). On the other hand, PDGFR SNP A13 was never detected in well

    differentiated tumors, whereas it was identified in 23% of moderately or poorly differentiated

    ones (p=0.053).

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    10/21

    Table 2Correlation of PDGFR A13 and B19 mutational status and clinical-pathologicalfeatures

    Clinical Features PDGFR exon 13 PDGFR exon 19

    WT SNP p WT SNP p

    N (%) N (%) N (%) N (%)

    Gender 0.754 0.346

    Female 24 (85.7) 4 (14.3) 10 (34.5) 19 (65.5)

    Male 36 (80.0) 9 (20.0) 23 (46.9) 26 (53.1)

    Age 0.127 0.653

    68-years old (median) 34 (89.5) 4 (10.5) 18 (45.0) 22 (55.0)

    > 68-years old (median) 26 (74.3) 9 (25.7) 15 (39.5) 23 (60.5)

    CEA (carcinoembryonic antigen) 1.000 0.813

    Within normal range 39 (83.0) 8 (17.0) 21 (41.2) 30 (58.8)

    ULN (5 ng/ml) 21 (80.8) 5 (19.2) 12 (44.4) 15 (55.6)

    Primary tumour location 1.000 0.051 Colon 47 (82.5) 10 (17.5) 22 (36.1) 39 (63.9)

    Rectum 13 (81.3) 3 (18.8) 11 (64.7) 6 (35.3)

    Tumour histology 0.401 0.221

    Conventional adenocarcinoma 52 (83.9) 10 (16.1) 30 (45.5) 36 (54.5)

    Mucinous or colloid adenocarcinoma 8 (72.7) 3 (27.3) 3 (25.0) 9 (75.0)

    TNM stage 0.196 0.170

    I-II 23 (92.0) 2 (8.0) 8 (30.8) 18 (69.2)

    III-IV 37 (78.7) 10 (21.3) 24 (47.1) 27 (52.9)

    Tumour differentiation 0.053 0.586 Well differentiated 15 (100) 0 (0.0) 7 (36.8) 12 (63.3)

    Moderately or poorly differentiated 37 (77.1) 11 (22.9) 23 (47.9) 25 (52.1)

    Surgery of primary tumor 0.578 0.389

    Yes 55 (80.9) 13 (19.1) 32 (43.8) 41 (56.2)

    No 5 (100.0) 0 (0.0) 1 (20.0) 4 (80.0)

    Surgery of metastasis 0.953 0.451

    Yes 13 (86.7) 2 (13.3) 9 (50.0) 9 (50.0)

    No 47 (81.0) 11 (19.0) 24 (40.0) 24 (40.0)

    Adjuvant chemotherapy 0.295 0.986

    Yes 28 (87.5) 4 (12.5) 14 (42.4) 19 (57.6)

    No 32 (78.0) 9 (22.0) 19 (42.2) 26 (57.8)

    Chemotherapy for advanced disease 0.683 0.929

    Yes 36 (83.7) 7 (16.3) 18 (41.9) 25 (58.1)

    No 24 (80.0) 6 (20.0) 15 (42.9) 20 (57.1)

    PDGFR and PDGFR genetic variants and colon cancer survival

    Overall survival of patients according to PDGFR-A13 and -B19 SNPs identified is depicted

    in Table 3. No significant impact in overall survival was observed for SNP A13. On the

    contrary, 5-year survival of patients PDGFR-B19 WT was substantially greater than thatobserved in those harboring the SNP (51% vs 17%; p=0.073) (Figure 2). Multivariate

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    11/21

    analyses showed the presence of the B19 SNP variant was a significant independent predictor

    of survival (HR 2.89, p=0.029). Other variable that retained independent prognostic value in

    the Cox regression model was TNM stage (pG,

    rs1873778; exon 13 (A13): c.1809G>A, rs10028020; and exon 17 (A17): c.2439+58C>A,

    rs2412559] and one in PDGFR [exon 19 (B19): c.2601A>G, rs246395]. SNP B19, present

    in 4 CRC cell lines (LS174T, LS180, SW48, COLO205) and in 58% of patients, had a

    substantial impact on overall survival, with 5-year survival rates of 51% for patients with

    PDGFR B19 wild type tumors versus 17% for those harboring the SNP variant (c.2601A>G).

    This is the first study to analyze the PDGFR genotype in a series of human colorectal cancer

    and its correlation with different clinicopathological features, and to demonstrate a significantassociation of a PDGFR SNP with patients outcome.

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    12/21

    Angiogenesis is a complex process controlled by a number of interconnected signaling

    pathways, among which PDGF and their receptors play a critical role. Moreover, PDGFR has

    been the target for many newly developed anticancer drugs, some of them with proven

    efficacy in CRC (i.e. regorafenib) [7] and some that have failed to demonstrate a benefit in

    patients with this tumor type (i.e. sunitinib, sorafenib) [16,17]. Despite this, however, only

    few studies have analyzed the clinical implications of PDGF/PDGFR expression in colorectalcancer. In this regard, Schimanski and cols reported that specific receptor tyrosine kinases

    (TK) were overexpressed in K-ras mutated CRC [22]. In particular, VEGFR1, VEGFR2 and

    PDGFR expression, documented in 95%, 46% and 62% of tested samples, respectively,

    were significantly linked to K-ras codon 12 or 13 mutations. Whether this could translate into

    a higher likelihood of responding to TK inhibitors, however, is a matter of speculation. On

    the other hand, Wheler et al. reported, in a series of 99 human colorectal carcinomas, that co-

    expression of PDGFR/, observed in 57% of tumor samples, was significantly associated

    with lymphatic metastasis (P=0.007) and advanced tumor stage (P=0.03) [23]. Similarly, high

    PDGFR tumor stromal expression significantly correlated with more aggressive clinical

    behavior in patients with breast cancer, including high histopathological grade, estrogen

    receptor negativity, high HER2 expression and shorter survival [24].

    Nevertheless, PDGFR genetic variants had never been previously assessed in CRC patients.In our study, four genetic variants were identified, all of them corresponding to SNPs

    previously reported in public databases. Three of them were silent mutations (A12, A13 and

    B19) and the other one was an intronic insertion (A17). PDGFR exon 12 SNP (rs1873778),

    present in homozygosis in all CRC cell lines and 100% of analyzed tumor samples, has been

    also described in other neoplasias although in a smaller proportion of patients, including KIT

    and FLT3 mutation-negative core binding factor (CBFL) acute myeloid leukemias (14% of

    35 patients) [25], cervical adenosquamous carcinomas (30% of 30 patients) [26] and gliomas

    (7% of 86 patients) [27]. In this last study, no association was found between the presence of

    this mutation and PDGFR tissue expression. Our results are in agreement with the

    distribution reported for a European Caucasian population at the NCBI website

    (http//:www.ncbi.nlm.nih.gov/sites/entrez/), being the G-allele the most frequently

    encountered (p=0.98). PDGFR exon 13 SNP (rs10028020), detected in heterozygosis in 2

    (LS174T and LS180) of the 8 cell lines examined and in 18% of tumor samples, was

    associated with poorer tumor differentiation but no significant correlation was found with

    survival. This polymorphism had been first reported also in heterozygosis by Trojani et al. in

    34% of CBFL acute leukemias [25], although potential association of this genotype with

    clinical features or patients outcome was not explored by these authors. Finally, neither

    PDGFR exon 17 SNP (rs2412559), identified in all of our patients, nor PDGFR exon 19

    SNP (rs246395), present in 58% of them, had been previously described in human cancers.PDGFR B19 SNP has been reported to be present in the general population with a frequency

    of 37%, and was more commonly encountered in our study population among colon primary

    tumors (64%) than in tumors of rectal origin (35%). Of note, and despite not being an

    activating mutation, the B19 SNP was found to be a significant prognostic factor (HR: 2.89,

    p=0.029) independent of tumor stage or patients age. This negative effect on patients

    survival did not differ according to primary tumor location (data not shown).

    That the identified SNP in exon 19 of PDGFR may indeed have relevant biological

    implications is further supported by the fact that analysis of protein content in cell lines

    demonstrated the presence of the B19 SNP clearly correlated with higher protein levels of the

    PDGF receptor , also in its phosphorylated state. PDGF pathway constitutive activationmaintains highly active MEK, thus phosphorylating Bad and inhibiting apoptosis [14,15].

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    13/21

    Increased PDGF pathway activation has been also shown to contribute to drug resistance by

    activating the PI3K pathway [14,15]. Whether or not the presence of this SNP may portend

    particular sensitivity to PDGFR-targeted agents is a matter of speculation but certainly

    deserves further investigation due to its relevant potential clinical applications.

    On the contrary, no relevant findings were identified in our series regarding VEGFR2 TKdomain SNP analysis. As in other solid tumors, overexpression of VEGF mRNA and protein

    has been associated with tumor progression and poor prognosis of colon carcinoma [28]. The

    VEGF-A gene is known to be highly polymorphic and harbors numerous SNPs, especially in

    the promoter, 5- and 3-untranslated regions (UTR), which contain key regulatory elements

    that are sensitive to hypoxia [29]. These SNPs contribute to the high variability in VEGF

    production among tissues and have been associated with cancer susceptibility, progression,

    and anti-VEGF therapeutic response in subjects with a variety of solid tumors including

    colorectal cancer. For example, the 936 T-allele has been associated with increased risk of

    CRC, advanced stage of disease and worse prognosis, whereas the 634 C allele was

    predictive of decreased risk and improved survival. SNPs have also been identified in the

    VEGF receptor genes, although the literature in this topic is still very sparse. Very recently,the VEGFR-1 319 C/A SNP, located in the promoter region of the gene, has been reported to

    be associated with response to therapy in a cohort of 218 CRC patients treated with differentbevacizumab-containing regimens [30]. In this study by Hansen et al., response rates were

    significantly higher in patients homozygous for the A-allele (AA) than in patients with the C-

    allele genotype (CC or CA) (56% vs 39%, p=0.015). Similar results were also documented in

    bevacizumab-treated pancreatic cancer patients [31]. In addition, functional relevance has

    been demonstrated for several SNPs in the VEGFR-1 and VEGFR-2 genes, particularly SNPs

    1192C/T (V2971I; rs2305948) and 1719T/A (H472Q; rs1870377). These SNPs are located in

    exons 7 and 11, and lead to amino acid changes potentially interfering with the receptors

    binding affinity to VEGF-A. In the current study, however, we aimed to explore potential

    genetic variations in the TK domain of the VEGFR-2 (exons 17 to 26), which would be

    expected to have relevant functional consequences. No mutations were however detected in

    our study population in these gene domains.

    Identification of relevant SNPs in critical genes involved in angiogenesis may therefore

    become valuable tools in assessing risk or predicting cancer response to therapy or prognosis.

    However, no consensus exists at present regarding the use of any of these for clinical

    decisions as many studies have reported diverging, conflicting or inconclusive results.

    Multiple reasons may be responsible for these discrepancies, including gender and interethnic

    differences in the distribution of alleles, heterogeneous study populations and small sample

    sizes, different sources of DNA (i.e., tumor vs germline) and different methods for SNPanalyses, lack of corrections for multiple testing, links to other loci in the gene or related

    genes responsible for the observed effect, bias due to post-transcriptional gene regulation, or

    simultaneous presence of somatic or epigenetic changes that may influence outcome.

    Prospective validation in appropriately sized and controlled studies is therefore required

    before these genetic variants may be used in clinical practice.

    Conclusion

    In conclusion, the present study has identified, for the first time, PDGFR genetic variants

    with relevant clinical and biological implications. In particular, the G-allele genotype of

    PDGFR exon 19 SNP (rs246395), which was commonly encountered in our series of CRC

    patients (58%), was associated with increased pathway activation and poorer survival.

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    14/21

    Further studies to assess the functional consequences of this genetic variant, as well as to

    validate its role as a prognostic marker in this disease are certainly warranted. Implications

    regarding its potential influence in response to PDGFR-targeted agents remain to be

    elucidated.

    Competing interest

    The authors declare no conflict of interest.

    Authors contributions

    Conception and design: RGC, ACM, LPA. Molecular genetic and protein analysis in human

    samples and cancer cell lines: ACM, JI, SMP, MDP, SM. Pathological assessment: AC, FLR.

    Collection and assembly of data: ACM, JI, ILC, SMP, MDP, SM, PEG, RGC. Data analysis

    and interpretation: RGC, LPA, ACM, JI, AC, PEG. Manuscript writing: PEG, SMP, AC,

    LPA, RGC. Final approval of manuscript: All authors.

    Novelty and impact of present research

    This study has identified, for the first time, PDGFR genetic variants with relevant clinical

    and biological implications in colorectal cancer. In particular, the G-allele genotype of

    PDGFR exon 19 SNP (rs246395), encountered in 58% of tumor samples from colorectal

    cancer patients, was associated with increased pathway activation and significantly poorer

    survival.

    Acknowledgements

    We would like to thank the technical staff from Bionostra for technical assistance. This work

    was supported by a grant of the Ministerio de Ciencia y Tecnologia of Spain (FIT-010000-

    2006-45). PEG is funded by a Rio Hortega grant (09/00207) from the Instituto de Salud

    Carlos III (ISCiii), Ministerio de Sanidad, Spain. SMP is funded by the ISCiii (CD1100153),

    Fundacin Cientfica de la Asociacion Espaola Contra el Cancer (AECC), Consejeria de

    SaludJunta de Andalucia (PI-0224/2009) and Fundacion Mutua Madrilea (2009). MDP is

    funded by the ISCiii (CD0900148). LPA is funded by the ISCiii (PI081156 and PI1102688),

    Consejera de Innovacion, Ciencia y EmpresaJunta de Andalucia (P08-CVI-04090) and the

    75th

    Anniversary Roche Spain Fellowship. RGC is funded by the ISCiii (PI 10.02164).

    References

    1. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM: GLOBOCAN 2008 v1.2,

    Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 10 [Internet]. Lyon,

    France: International Agency for Research on Cancer; 2010. http://globocan.iarc.fr, accessed

    on 31/08/2011.

    2. Vieitez JM, Garca-Carbonero R, Aparicio J, Feliu J, Gonzlez-Flores E, Grande E, Prez-

    Hoyos T, Salud A, Torres E, Valero M, Valladares-Ayerbes M, Daz-Rubio E:

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    15/21

    Recommendations and expert opinion on the adjuvant treatment of colon cncer inSpain.Clin Transl Oncol 2011, 13(11):798804.

    3. Garca-Carbonero R, Gmez Espaa MA, Casado Senz E, Alonso Ordua V, Cervantes

    Ruiprez A, Gallego Plazas J, Garca Alfonso P, Juez Martel I, Gonzlez Flores E, Lomas

    Garrido M, Isla Casado D: SEOM clinical guidelines for the treatment of advancedcolorectal cancer.Clin Transl Oncol 2010, 12(11):729734.

    4. Aranda E, Abad A, Carrato A, Cervantes A, Garca-Foncillas J, Garca Alfonso P, Garca

    Carbonero R, Gmez Espaa A, Tabernero JM, Daz-Rubio E: Treatment

    recommendations for metastatic colorectal cancer. Clin Transl Oncol 2011, 13(3):162

    178.

    5. Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, Berlin J,

    Baron A, Griffing S, Holmgren E, Ferrara N, Fyfe G, Rogers B, Ross R, Kabbinavar F:

    Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal

    cancer.N Engl J Med2004, 350(23):23352342.

    6. Joulain F, Van Cutsem E, Usman I, Hoyle M, Allegra C: Aflibercept versus placebo in

    combination with FOLFIRI in previously treated metastatic colorectal cancer (mCRC):Mean overall survival (OS) estimation from a phase III trial (VELOUR).J Clin Oncol

    2012, 30:s3602.

    7. Van Cutsem E, Sobrero AF, Siena S, Falcone A, Ychou M, Humblet Y, Bouche O, Mineur

    L, Barone C, Adenis A, Tabernero J, Yoshino T, Lenz HJ, Goldberg RM, Sargent DJ, Cihon

    F, Wagner A, Laurent D, Grothey A, on behalf of CORRECT Investigators: Phase IIICORRECT trial of regorafenib in metastatic colorectal cancer (mCRC).J Clin Oncol2012, 30(suppl; abstr 3502).

    8. Carmeliet P, Jain RK: Angiogenesis in cancer and other diseases.Nature 2000, 407:249

    257.

    9. Weis SM, Cheresh DA: Pathophysiological consequences of VEGF-induced vascular

    permeability.Nature 2005, 437:497504.

    10. Sakurai T, Kudo M: Signaling pathways governing tumor angiogenesis. Oncology

    2011, 81(Suppl 1):2429.

    11. Arora A, Scholar EM: Role of tyrosine kinase inhibitors in cancer therapy.JPharmacol Exp Ther2005, 315:971979.

    12. Ivy SP, Wick JY, Kaufman BM: An overview of small-molecule inhibitors of VEGFRsignaling.Nature Rev Clin Oncol 2009, 6:569579.

    13. Hoch RV, Soriano P: Roles of PDGF in animal development.Development 2003,

    130:47694784.

    14. Yu J, Ustach C, Kim HR: Platelet-derived growth factor signaling and human cancer.

    J Biochem Mol Bio 2003, 36(1):4959.

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    16/21

    15. Andrae J, Gallini R, Betsholtz C: Role of platelet-derived growth factors in physiology

    and medicine.Genes Dev 2008, 22(10):12761312.

    16. Carrato A, Swieboda-Sadlej S-S, Lim R, Roman L, Shparyk Y, Bondarenko I, Lin X,

    Lechuga MJ, Tursi JM, Sun Y, De la Cruz JA, Jonker D, Van Cutsem E: Final results from

    a randomized, double-blind, phase III study of sunitinib plus FOLFIRI vs placebo plusFOLFIRI in first-line treatment of patients (pts) with metastatic colorectal cancer . InProceedings of the 12

    th World Cancer on Gastrointestinal Cancer. Barcelona, Spain:;

    2010:O-0026. 30 June-3 July.

    17. Tabernero J, Garcia-Carbonero R, Khne CH, et al: A phase 2b, double-blind,

    randomized study evaluating the efficacy and safety of sorafenib (SOR) compared with

    placebo (PBO) when administered in combination with chemotherapy (modified

    FOLFOX6) for first-line treatment (tx) of patients (pts) with metastatic colorectalcancer (mCRC), the RESPECT trial. In Proceeding of the 2011 European

    Multidisciplinary Cancer Congress. Stockholm, Sweden: Abstract LBA19; 2011. 2327

    September.

    18. In AJCC Cancer Staging Manual. 7th edition. Edited by Edge SB, Byrd DR, Compton

    CC, Fritz AG, Greene FL, Trotti A.: Springer; 2010.

    19. Kaplan EL, Meier P: Non parametric estimation from incomplete observations.J AmStat Soc A. 1972, 35:185206.

    20. Peto R, Peto J: Asymptotically efficient rank invariant tests procedures.J R Stat Soc A

    1972, 135(2):185207.

    21. Cox DR: Regression models and life tables.J R Stat Soc B 1972, 34:187220.

    22. Schimanski CC, Zimmermann T, Schmidtmann I, Gockel I, Lang H, Galle PR, Moehler

    M, Berger MR: K-ras mutation status correlates with the expression of VEGFR1,

    VEGFR2, and PDGFRalpha in colorectal cancer.Int J Colorectal Dis 2010, 25(2):181

    186.

    23. Wehler TC, Frerichs K, Graf C, Drescher D, Schimanski K, Biesterfeld S, Berger MR,

    Kanzler S, Junginger T, Galle PR, Moehler M, Gockel I, Schimanski CC:

    PDGFRalpha/beta expression correlates with the metastatic behavior of human

    colorectal cancer: a possible rationale for a molecular targeting strategy. Oncol Rep2008, 19(3):697704.

    24. Paulsson J, Sjblom T, Micke P, Pontn F, Landberg G, Heldin CH, Bergh J, Brennan

    DJ, Jirstrm K, Ostman A: Prognostic significance of stromal platelet-derived growthfactor beta-receptor expression in human breast cancer.Am J Pathol 2009, 175(1):334

    341.

    25. Trojani A, Ripamonti CB, Penco S, Beghini A, Nadali G, Di Bona E, Viola A, Castagnola

    C, Colapietro P, Grillo G, Pezzetti L, Ravelli E, Patrosso MC, Marocchi A, Cuneo A, Ferrara

    F, Lazzarino M, Pizzolo G, Cairoli R, Morra E: Molecular analysis of PDGFRA and

    PDGFRB genes by rapid single-strand conformation polymorphism (SSCP) in patients

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    17/21

    with core-binding factor leukaemias with KIT or FLT3 mutation.Anticancer Res 2008,

    28(5A):27452751.

    26. Longatto-Filho A, Pinheiro C, Martinho O, Moreira MA, Ribeiro LF, Queiroz GS,

    Schmitt FC, Baltazar F, Reis RM: Molecular characterization of EGFR, PDGFRA and

    VEGFR2 in cervical adenosquamous carcinoma.BMC Cancer2009, 9:212.

    27. Martinho O, Longatto-Filho A, Lambros MB, Martins A, Pinheiro C, Silva A, Pardal F,

    Amorim J, Mackay A, Milanezi F, Tamber N, Fenwick K, Ashworth A, Reis-Filho JS, Lopes

    JM, Reis RM: Expression, mutation and copy number analysis of platelet-derived

    growth factor receptor A (PDGFRA) and its ligand PDGFA in gliomas. Br J Cancer

    2009, 101(6):973982.

    28. Hansen TF, Jakobsen A: Clinical implications of genetic variations in the VEGFsystem in relation to colorectal cancer.Pharmacogenomics. 2011, 12(12):16811693.

    29. Jain L, Vargo CA, Danesi R, Sissung TM, Price DK, Venzon D, Venitz J, Figg WD: Therole of vascular endothelial growth factor SNPs as predictive and prognostic markersfor major solid tumors.Mol Cancer Ther2009, 8:24962508.

    30. Hansen TF, Christensen RD, Andersen RF, Spindler KL, Johnsson A, Jakobsen A: The

    predictive value of single nucleotide polymorphisms in the VEGF system to the efficacy

    of first-line treatment with bevacizumab plus chemotherapy in patients with metastaticcolorectal cancer: Results from the Nordic ACT trial. Int J Colorectal Dis 2012,27(6):715720.

    31. Lambrechts D, Claes B, Delmar P, Reumers J, Mazzone M, Yesilyurt BT, Devlieger R,

    Verslype C, Tejpar S, Wildiers H, de Haas S, Carmeliet P, Scherer SJ, Van Cutsem E: VEGF

    pathway genetic variants as biomarkers of treatment outcome with bevacizumab: ananalysis of data from the AViTA and AVOREN randomised trials. Lancet Oncol 2012,13(7):724733.

    Additional file

    Additional_file_1 as DOCAdditional file 1 Supplementary Tables

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    18/21

    Figure 1

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    19/21

    Time (months)

    12096724824

    1,0

    0,8

    0,6

    0,4

    0,2

    0,0

    SNP-censoredWT-censoredSNPWT

    PDGFR B19

    HR=1.93P=0.073

    Overall Survival by PDGFR exon 19 Genotype

    Figure 2

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    20/21

    Figure 3

  • 7/30/2019 PDGFRalpha/beta and VEGFR2 polymorphisms in colorectal cancer: incidence and implications in clinical outcome

    21/21

    Additional files provided with this submission:

    Additional file 1: 1477341248753074_add1.doc, 70Khttp://www.biomedcentral.com/imedia/3008989598471490/supp1.doc

    http://www.biomedcentral.com/imedia/3008989598471490/supp1.dochttp://www.biomedcentral.com/imedia/3008989598471490/supp1.doc